Skip to main content

Impact of placental malaria on maternal, placental and fetal cord responses and its role in pregnancy outcomes in women from Blue Nile State, Sudan

Abstract

Background

The sequestration of Plasmodium falciparum infected cells in the placenta results in placental malaria (PM). It activates the mother's immune cells and induces secretion of inflammatory cytokines, which might influence pregnancy outcomes. This study aims to investigate the cytokines (levels IL-4, IL-6, IL-10, IL-17A, and INF γ) in maternal peripheral, placental, and umbilical cord blood in response to PM and the extent to which this may influence maternal haemoglobin levels and birth weight.

Methods

A total of 185 consenting Sudanese women from Blue Nile State were enrolled at delivery time in a cross-sectional study conducted between Jan 2012-Dec 2015. Malaria infection in the collected maternal peripheral, placental, umbilical cord samples was determined microscopically, and ELISA was used to measure the plasma levels IL-4, IL-6, IL-10, IL-17A, and INF γ in the collected positive and negative malaria samples.

Results

Elevated levels of IL-4 and IL-10 and reduced levels of IL-6 were detected in the malaria positive samples in comparison to the negative ones in the three types of the samples investigated. Maternal, IL-4 and IL-10 were significantly higher in the samples collected from the PM infected group compared to the non-infected control (P < 0.001). While the absence of PM was significantly associated with the IL-6 and maternal IFN-γ levels, maternal IL-17A, placental and umbilical cord IFN-γ levels showed no significant difference (P = 0.214, P = 0.065, P = 0.536, respectively) due to infection. Haemoglobin level and birth weight were increased in the group with high levels of IL-6 and IL-17A, but not in the group with IL-4 and IL-10 levels. While significantly negative correlation was found between IFN-γ levels and birth weight for all three types of samples, only maternal peripheral IFN-γ level was significantly positively correlated with maternal haemoglobin (r = 0.171, P = 0.020).

Conclusion

These results suggest that PM induces mother’s immune response and impairs her cytokine profile, which might alter maternal haemoglobin levels and the baby's birth weight.

Background

Plasmodium falciparum infections during pregnancy result in pregnancy-associated malaria (PAM) and placental malaria (PM). In PM, the parasite ligand VAR2CSA mediates adhesion of infected erythrocytes (IEs) to chondroitin sulphate A CSA in the placental syncytiotrophoblast [1, 2]. Accumulation of IES in the placenta induces pathological changes that alter the materno-fetal exchange system and can lead to maternal morbidity and severe fetal and neonatal complications [3, 4]. While pregnancy is characterized by an induced immunosuppression [5], an activated immune system during pregnancy might have a role in protection against malaria and poor delivery outcomes [6, 7], and it might decrease the risk of both P. falciparum and Plasmodium vivax infections [8].

It is also known that P. falciparum IEs induce inflammation by monocytic infiltration of the malaria infected placenta, which is associated with maternal anaemia and low birth weight (LBW) [9, 10]. This inflammation may influence cellular functions by altering the balance of cytokines and chemokines in the peripheral and placental blood of the women [11, 12], and some cytokines can help resolve infections while others may also contribute to pathology [13,14,15]. Although the balance between pro- and anti-inflammatory cytokines is a key factor in the regulation of an effective immune response to PM [16], the roles of the produced cytokines are controversial. Among the cytokines that are produced, IL-27 and IL-6 can elicit both pro-inflammatory and anti-inflammatory effects, and high concentrations of IL-6 may protect against PM [17].

IFN-γ and IL-17A levels have been found to be negatively correlated with PM infection and maternal anaemia [15], and an association of IFN-γ with protection from malaria has been reported [18, 19]. Increased Th1 response due to infection was found to be incompatible with a successful pregnancy in mice [20]. An elevated level of IFN-γ may result in trophoblast damage, preterm delivery, and LBW associated with PM [21,22,23]. IL-17A may facilitate local inflammation by recruiting and activating immune cells, leading to the upregulation of inflammatory cytokine production [24], but little is known about its role in malaria infection during pregnancy and PM.

Plasmodium falciparum infection during pregnancy increases the placental levels of IL-10 and IL-4 [13, 25]. Elevated levels of the anti-inflammatory cytokine IL-10 in placental plasma were associated with PM, and were implicated in the pathogenesis of severe anaemia [9, 12, 13]. Moreover, IL-10 has been identified as an immunosuppressive cytokine associated with poor pregnancy outcome in a mouse model of PM [26]. Furthermore, placental infection influences fetal immune responses by affecting the Th1/Th2 balance in umbilical cord blood through IL-10 production by T regulatory cells as a result of infection [27].

An understanding of the inflammatory responses in the mother and foetus and the cause of LBW during PM could help optimize efforts to prevent the consequence of placental sequestration that results in poor birth outcomes.

This study aims to investigate the inflammatory environment in maternal peripheral, placental, and umbilical cord blood in response to PM and the extent to which this may influence pregnancy outcomes. The study was conducted with a cohort of Sudanese women from the Blue Nile State, which has a high rate of seasonal malaria transmission.

Methods

Study area and population

This cross-sectional study consisted of a cohort of pregnant women in Blue Nile State, Sudan at their time of delivery, who were recruited between January 2012 and December 2015. The study area, population, and the sample collection method have been described elsewhere [28]. Briefly, a total of 1149 consenting pregnant women were recruited for assessment of the prevalence and risk factors of PM in the study area. The study participants for the sub-study described in this paper included a group of 185 women for whom at least 2 ml of peripheral, placental and cord plasma were available and they signed additional consent for the immune response investigation. Information on the mother's socio-demographic characteristics, such as age, parity, education, use of bed nets and anti-malarial drugs, was obtained via a questionnaire. The haemoglobin level was assessed, and maternal anaemia at delivery was defined as a haemoglobin level < 11 g/d. The neonates were weighed immediately after birth.

Sample collection

Maternal peripheral, placental and umbilical cord blood samples were collected immediately after delivery in heparinized vacutainer tubes. A portion of each sample was used to prepare smears for malaria microscopy and to determine the haemoglobin levels in maternal blood. The rest of the blood was centrifuged at 3000 g for 3 min, and the undiluted plasma was aliquoted into several tubes (to avoid freeze and thaw) and stored at −80 °C until thawed on the day that cytokine assays were performed. The blood smears were routinely stained with Giemsa and microscopically examined by two expert microscopists to determine the presence of malaria parasites.

Measurement of the cytokines

The plasma of maternal peripheral, placental and neonate umbilical cord blood was simultaneously subjected to cytokine screening of IL-4, IL-6, IL-10, IL-17A and INF γ using Human ELISA MAX™ Deluxe commercial kits (BioLegend, USA) according to the manufacturer's instructions. The sensitivity of the assay for each cytokine was 2 pg/ml, which was the minimum detectable concentration of each cytokine.

Statistical analysis

Data were analysed using SPSS software for Windows. Continuous data were checked for normality using the Shapiro–Wilk test; they were expressed as median (interquartile) if they were found to not be normally distributed. The Mann–Whitney U test and the Kruskal–Wallis H test (non-parametric) were used to compare the non-normally distributed continuous variables between two and three groups, respectively. Spearman's correlation test was used to assess the correlations between the non-normally distributed continuous variables.

Results

General characteristics

A total of 185 samples were collected at delivery time from maternal peripheral, placental, and umbilical cord blood. The median (interquartile) of the age was 20 (18−22 years). The median (interquartile) of the parity was 2 (1−3). The median (interquartile) of the haemoglobin level of the participants was 10.2 (9.3−11.3) g/dl. A total of 127 (68.6%) women had anaemia (haemoglobin < 11.0 g/dl). Ninety-two (49.7%) of the samples from the maternal peripheral blood, 97(52.4%) of the placental and 55 (29.7%), the umbilical cord was blood film positive for P. falciparum infection. Forty-three (23.2%) of the investigated blood films were positive in the three compartments of the same patient.

The median (interquartile) of the birth weight was 2.4 kg (2.3−2.7). Ninety-eight (53.0%) of the new-borns had LBW (birth weight < 2.5 kg).

Cytokines levels in the maternal, placental and umbilical cord samples

With the exception of IL-17A levels, the cytokines levels were significantly lower in the umbilical cord samples than the maternal peripheral and placental samples. IL-17A level was similar in the maternal and umbilical cord, however the maternal and umbilical cord level was significantly lower compared with the placental level (Table 1).

Table 1 Comparisons of median cytokine levels (interquartile range) in the maternal, placental and cord blood compartments of women positive and negative for malaria infections

Cytokines levels and malaria infections

While the levels (in three compartments) of IL- 4 and IL-10 were significantly higher, the levels of IL-6 in all the compartments and maternal IFN-γ were significantly lower in the PM positive samples in comparison to the PM negative samples. Maternal peripheral IL-17A levels and the placental and umbilical IFN-γ levels showed no significant difference between the PM infected group and the non-infected group (Table 1).

Cytokines levels and parity

There was no significant difference in the maternal peripheral and placental IL-17A levels between the primiparous women and the parous women. The level of IL-4 was significantly higher in the primiparous women in all three types of samples (maternal peripheral, placental, and umbilical cord blood). The IL-6 levels in the maternal peripheral, placental and umbilical cord blood, and the umbilical cord levels of IL-17A were significantly lower in the primiparous women. The maternal peripheral and placental IL-10 levels and the placental and umbilical cord IFN-γ levels (no significant difference in the maternal peripheral IFN-γ levels) were significantly higher in the primiparous women in comparison to the parous women (Table 2).

Table 2 Comparison of cytokines level between primiparous and parous women

Correlations between cytokines

The IL-4 levels in the plasma of the three types of blood samples were significantly negatively correlated with the IL-6 levels, and they were positively correlated with the IL-10 levels. There was a borderline negative correlation between the maternal peripheral IL-4 levels and the maternal peripheral IFN-γ levels. A negative correlation and a positive correlation were found between the placental IL-4 levels and the placental IL-17A levels and the IFN-γ level, respectively. In the umbilical cord samples, the level of IL-4 was negatively correlated with the level of IL-17A.

The IL-6 levels in all three types of the blood samples were significantly negatively correlated with the levels of IL-10 and positively correlated with the levels of IL-17A. The maternal and placenta IL-6 levels were positively and negatively, respectively correlated with maternal and placenta IFN-γ.

While no correlation was found between the maternal peripheral level of IL-10 and the maternal peripheral levels of IL-17A and IFN-γ, negative correlations were found between the placental and umbilical cord levels of IL-10 and IL-17A, and a positive correlation was found between IL-10 levels and the level of placental and umbilical cord of IFN-γ. No correlation was found between the level of IL-17A for the maternal peripheral, placental and umbilical cord blood samples and the IFN-γ levels of the plasma in the three types of samples.

Correlations between cytokines, maternal haemoglobin and birthweight

Maternal haemoglobin was significantly negatively correlated with the levels of IL-4 and IL-10 in the maternal peripheral, placental and umbilical cord blood samples; however, no significant correlation was found between maternal haemoglobin and the level of the peripheral IL-17A. Significantly positive correlation and negative correlations were found between the levels of IL-6 and IFN-γ, respectively, and birth weight for all three types of samples, which was significantly negatively correlated with the levels of, IL-4 and IL-10. While no significant correlation was found between birth weight and maternal levels of IL-17A, it was significantly positively correlated with the levels of IL-17A in the placental and umbilical cord samples (Tables 3, 4, 5).

Table 3 Correlations between maternal plasma levels of cytokines, maternal hemoglobin and birth weight
Table 4 Correlations between placental plasma levels of cytokines, maternal hemoglobin and birth weight
Table 5 Correlations between umbilical cord plasma levels of cytokines, maternal hemoglobin and birth weight

Discussion

The present study found that malaria infection altered the investigated cytokines (IL-4, IL-6, IL-10, IL-17A and INF-γ) in Sudanese maternal, placental and neonatal plasma collected from Blue Nile State. Disturbance of cytokines secretions has been linked to many pathological disorders including poor pregnancy outcomes [15].

In the current study, the levels of IL-4 and IL-10 were elevated but the levels of IL-6 and IL-17 were reduced in the plasma of the maternal peripheral, placental and umbilical cord blood samples of the PM infected mothers in comparison to the non-infected group. Moreover, the maternal peripheral level of IL-17A and the placental and neonate IFN-γ levels were not significantly different due to the infection.

Several previous studies have shown elevated levels of type 2 anti-inflammatory cytokines IL-10 and IL-4 in the malaria-infected group [12, 13, 15, 23, 25, 29, 30]. Conversely, the present results were different from the previous findings in an area with unusable malaria transmission in Sudan, where higher levels of IL-4 and IL-10 in the non-infected group was reported [11]. The findings of the increased IL-6 levels in non-infected women is in agreement with the results of other studies [11, 16, 31, 32].

Interestingly, the level of cytokines in the maternal peripheral samples among the investigated groups was significantly correlated with the levels in the placental and umbilical cord samples. Nevertheless, plasma from the umbilical cord (except for the maternal peripheral IL-17A levels) contained significantly lower concentrations of the investigated cytokines in comparison to the peripheral and placental plasma in both study groups (PM infected and non-infected groups). Kabyemela et al. [25], in Tanzania, and Ibitokou [31], in Benin, reported similar levels of IFN-γ between the investigated groups. The results of the present study corroborate the findings of a previous study that reported higher levels of IFN-γ in infected placentas [9], and a study from Cameron that found an increase in maternal IFN-γ and IL-17-A levels in non-infected controls [15]. The elevated detection levels of IFN-γ in the investigated samples may be due to the ability of the innate immune response to produce IFN-γ to clear the parasite, probably as the first line of defense against both peripheral and placental infection [18]. It has been suggested that the differences in cytokine responses associated with malaria infection have a gravidity-based pattern [22].

Infiltration of immune cells in the placental intervillous spaces, due to the sequestration of P falciparum IEs, disrupting Th1 and Th2 cytokines balance in both placental and peripheral blood [22, 33]. Contradictory findings about the role of impaired cellular immune responses directed against malaria parasites in pregnancy and pathogenesis of PM have been described [12, 13, 15, 17, 21, 25, 34, 35]. However, the differences in the cytokine profiles between these studies may be attributed to variations in the cytokine measurement methods used and the malaria endemicity in different study areas, as well as differences in the diagnostic techniques and the study population.

In the present study, among the cytokines assessed, IL-4 and IL-10 were elevated and significantly correlated (P < 0.001) in the plasma of the three types of blood samples that were investigated; no correlation was found between the maternal peripheral levels of IL-10 and IFN-γ (P = 0.448). IL-17A was positively correlated with IL-6, but no correlation was found between the IFN-γ in the plasma of the three types of samples. IFN-γ and IL-17-A are both produced by effector helper T cells, and IL-17A can facilitate regulation of inflammatory cytokine production by accelerating specific inflammation via the recruitment and activation of immune cells. IL-6 is both a target of IL-17 and a differentiation factor for Th17 cells that can led to an increase in IL-17 [36]. Agudelo et al. [23] observed a significant correlation between IL-4 and IL-10 in the placental samples, but not in the peripheral blood, and levels of maternal IL-10 and IFN-γ were positively correlated. Moreover, Agudelo et al. [23] reported high expression of IFN-γ, TNF and IL-10 in the placental tissues and peripheral blood samples, and placental IL-4 in the infected women in comparison to the non-infected group. Due to the high expression associated with PM infection, it has been suggested that the IL-10 level in peripheral blood to be used as biomarker of placental inflammation related to PM [25] or as an immunosuppressive factor by decreasing anti-parasitic cellular immune responses [26].

The present study found that IL-6 and IFN-γ were associated birth weight and the maternal haemoglobin level, accompanied by the negative correlation of IL-4 and IL-10 in the plasma of the three types of samples investigated. It has been documented that there is a disturbance of cytokine equilibrium in malaria during pregnancy, and PM may be involved in many pathological disorders; it may also have negative consequences, such as LBW and reduced maternal haemoglobin level [37,38,39].

While the levels of IL-17A in the placental and umbilical cord samples were significantly positively correlated with birth weight and maternal haemoglobin, no significant correlation was found with the IL-17A levels in the maternal peripheral samples. The roles of IFN-γ and IL-10 in the malaria-infected women with maternal anaemia and baby birth weight was controversially documented in a reviewed by Seitz et al. [39]. Although Djontu et al. [16] reported no significant association between IL-6 level, maternal haemoglobin and baby birth weight, an elevated level of IL-6 was associated with anaemia in another study [29].

Similar to the current study’s findings, an association was reported between maternal haemoglobin and IL-17A levels and the peripheral plasma level of IFN-γ. It has been suggested that both cytokines provide protection against infection [15]. Furthermore, elevated levels of IL-17 with high levels of IL-4, IL-12 and IFN-γ were associated with haemoglobin loss in malaria recovered semi-immune mice [40]. Fitri et al. [41] reported that an imbalance between IL-17 and IL- 10 caused low fetal weight in Plasmodium berghei infection in mice.

Disturbance of pro-inflammatory cytokines and the inflammatory disorder of iron haemostasis led to the development of malarial anaemia [42]. Elevated levels of circulating IL-6, which play a vital role in T cells differentiation and immune response polarization, have been strongly related to reduced haemoglobin concentration in reticulocytes.

A limitation of the current study is that it did not check for the presence of other infections which could influence variations in cytokine levels as the study was conducted in a setting of availability of other common infections during pregnancy.

Conclusion

In the present study, maternal peripheral infection and PM induces mother’s immune response accompanied by secretion of various cytokines in the maternal peripheral, placental and umbilical cord blood in Sudanese women. The present findings support the evidence reported in previous studies, which found that PM affects cytokines levels in infected women. However, longitudinal studies are needed to understand the maternal immune response throughout the entire course of pregnancy.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

References

  1. Salanti A, Staalsoe T, Lavstsen T, Jensen AT, Sowa MP, Arnot DE, et al. Selective upregulation of a single distinctly structured var gene in chondroitin sulphate A-adhering Plasmodium falciparum involved in pregnancy-associated malaria. Mol Microbiol. 2003;49:179–91.

    Article  CAS  PubMed  Google Scholar 

  2. Salanti A, Dahlback M, Turner L, Nielsen MA, Barfod L, Magistrado P, et al. Evidence for the involvement of VAR2CSA in pregnancy-associated malaria. J Exp Med. 2004;200:1197–203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Beeson JG, Scoullar MJ, Boeuf P. Combating low birth weight due to malaria infection in pregnancy. Sci Transl Med. 2018;10:eaat1506.

    Article  PubMed  CAS  Google Scholar 

  4. Dellicour S, Tatem AJ, Guerra CA, Snow RW, ter Kuile FO. Quantifying the number of pregnancies at risk of malaria in 2007: a demographic study. PLoS Med. 2010;7:e1000221.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Fievet N, Cot M, Chougnet C, Maubert B, Bickii J, Dubois B, et al. Malaria and pregnancy in Cameroonian primigravidae: humoral and cellular immune responses to Plasmodium falciparum blood-stage antigens. Am J Trop Med Hyg. 1995;53:612–7.

    Article  CAS  PubMed  Google Scholar 

  6. Requena P, Barrios D, Robinson LJ, Samol P, Umbers AJ, Wangnapi R, et al. Proinflammatory responses and higher IL-10 production by T cells correlate with protection against malaria during pregnancy and delivery outcomes. J Immunol. 2015;194:3275–85.

    Article  CAS  PubMed  Google Scholar 

  7. Mens PF, Bojtor EC, Schallig HD. Molecular interactions in the placenta during malaria infection. Eur J Obstet Gynecol Reprod Biol. 2010;152:126–32.

    Article  CAS  PubMed  Google Scholar 

  8. McLean AR, Ataide R, Simpson JA, Beeson JG, Fowkes FJ. Malaria and immunity during pregnancy and postpartum: a tale of two species. Parasitology. 2015;142:999–1015.

    Article  CAS  PubMed  Google Scholar 

  9. Rogerson SJ, Pollina E, Getachew A, Tadesse E, Lema VM, Molyneux ME. Placental monocyte infiltrates in response to Plasmodium falciparum malaria infection and their association with adverse pregnancy outcomes. Am J Trop Med Hyg. 2003;68:115–9.

    Article  PubMed  Google Scholar 

  10. Mackintosh CL, Beeson JG, Marsh K. Clinical features and pathogenesis of severe malaria. Trends Parasitol. 2004;20:597–603.

    Article  CAS  PubMed  Google Scholar 

  11. Bayoumi NK, Bakhet KH, Mohmmed AA, Eltom AM, Elbashir MI, Mavoungou E, et al. Cytokine profiles in peripheral, placental and cord blood in an area of unstable malaria transmission in eastern Sudan. J Trop Pediatr. 2009;55:233–7.

    Article  PubMed  Google Scholar 

  12. Chêne A, Briand V, Ibitokou S, Dechavanne S, Massougbodji A, Deloron P, et al. Placental cytokine and chemokine profiles reflect pregnancy outcomes in women exposed to Plasmodium falciparum infection. Infect Immun. 2014;82:3783–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Suguitan AL Jr, Leke RG, Fouda G, Zhou A, Thuita L, Metenou S, et al. Changes in the levels of chemokines and cytokines in the placentas of women with Plasmodium falciparum malaria. J Infect Dis. 2003;188:1074–82.

    Article  CAS  PubMed  Google Scholar 

  14. Brabin B, Romagosa C, Abdelgalil S, Menendez C, Verhoeff FH, McGready R, et al. The sick placenta—the role of malaria. Placenta. 2004;25:359–78.

    Article  CAS  PubMed  Google Scholar 

  15. Megnekou R, Lissom A, Bigoga JD, Djontu JC. Effects of pregnancy-associated malaria on t cell cytokines in Cameroonian women. Scand J Immunol. 2015;81:508–14.

    Article  CAS  PubMed  Google Scholar 

  16. Djontu JC, Siewe SS, Edene YDM, Nana BC, Foko EVC, Bigoga JD, et al. Impact of placental Plasmodium falciparum malaria infection on the Cameroonian maternal and neonate’s plasma levels of some cytokines known to regulate T cells differentiation and function. Malar J. 2016;15:561.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Dobaño C, Berthoud T, Manaca MN, Nhabomba A, Guinovart C, Aguilar R, et al. High production of pro-inflammatory cytokines by maternal blood mononuclear cells is associated with reduced maternal malaria but increased cord blood infection. Malar J. 2018;17:177.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Moore JM, Nahlen BL, Misore A, Lal AA, Udhayakumar V. Immunity to placental malaria. I. Elevated production of interferon-γ by placental blood mononuclear cells is associated with protection in an area with high transmission of malaria. J Infect Dis. 1999;179:1218–25.

    Article  CAS  PubMed  Google Scholar 

  19. Othoro C, Moore JM, Wannemuehler KA, Moses S, Lal A, Otieno J, et al. Elevated gamma interferon-producing NK cells, CD45RO memory-like T cells, and CD4 T cells are associated with protection against malaria infection in pregnancy. Infect Immun. 2008;76:1678–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Raghupathy R. Th1-type immunity is incompatible with successful pregnancy. Immunol Today. 1997;18:478–82.

    Article  CAS  PubMed  Google Scholar 

  21. Fried M, Muga RO, Misore AO, Duffy PE. Malaria elicits type 1 cytokines in the human placenta: IFN-gamma and TNF-alpha associated with pregnancy outcomes. J Immunol. 1998;160:2523–30.

    CAS  PubMed  Google Scholar 

  22. Rogerson SJ, Hviid L, Duffy PE, Leke RF, Taylor DW. Malaria in pregnancy: pathogenesis and immunity. Lancet Infect Dis. 2007;7:105–17.

    Article  CAS  PubMed  Google Scholar 

  23. Agudelo OM, Aristizabal BH, Yanow SK, Arango E, Carmona-Fonseca J, Maestre A. Submicroscopic infection of placenta by Plasmodium produces Th1/Th2 cytokine imbalance, inflammation and hypoxia in women from north-west Colombia. Malar J. 2014;13:122.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24:677–88.

    Article  CAS  PubMed  Google Scholar 

  25. Kabyemela ER, Muehlenbachs A, Fried M, Kurtis JD, Mutabingwa TK, Duffy PE. Maternal peripheral blood level of IL-10 as a marker for inflammatory placental malaria. Malar J. 2008;7:26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Megnekou R, Staalsoe T, Hviid L. Cytokine response to pregnancy-associated recrudescence of Plasmodium berghei infection in mice with pre-existing immunity to malaria. Malar J. 2013;12:387.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Bisseye C, Van Der Sande M, Morgan W, Holder A, Pinder M, Ismaili J. Plasmodium falciparum infection of the placenta impacts on the T helper type 1 (Th1)/Th2 balance of neonatal T cells through CD4+ CD25+ forkhead box P3+ regulatory T cells and interleukin-10. Clin Exp Immunol. 2009;158:287–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Omer SA, Idress HE, Adam I, Abdelrahim M, Noureldein AN, Abdelrazig AM, et al. Placental malaria and its effect on pregnancy outcomes in Sudanese women from Blue Nile State. Malar J. 2017;16:374.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Chandrasiri UP, Randall LM, Saad AA, Bashir AM, Rogerson SJ, Adam I. Low antibody levels to pregnancy-specific malaria antigens and heightened cytokine responses associated with severe malaria in pregnancy. J Infect Dis. 2014;209:1408–17.

    Article  CAS  PubMed  Google Scholar 

  30. Okamgba OC, Ifeanyichukwu MO, Ilesanmi AO, Chigbu LN. Variations in the leukocyte and cytokine profiles between placental and maternal circulation in pregnancy-associated malaria. Res Rep Trop Med. 2018;9:1–8.

    PubMed  PubMed Central  Google Scholar 

  31. Ibitokou SA, Boström S, Brutus L, Ndam NT, Vianou B, Agbowaï C, et al. Submicroscopic infections with Plasmodium falciparum during pregnancy and their association with circulating cytokine, chemokine, and cellular profiles. Clin Vaccine Immunol. 2014;21:859–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Lima FA, Barateiro A, Dombrowski JG, de Souza RM, Costa DS, Murillo O, et al. Plasmodium falciparum infection dysregulates placental autophagy. PLoS ONE. 2019;14:e0226117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fievet N, Moussa M, Tami G, Maubert B, Cot M, Deloron P, et al. Plasmodium falciparum induces a Th1/Th2 disequilibrium, favoring the Th1-type pathway, in the human placenta. J Infect Dis. 2001;183:1530–4.

    Article  CAS  PubMed  Google Scholar 

  34. Achidi EA, Apinjoh TO, Titanji VP. Malaria parasitemia and systemic cytokine bias in pregnancy. Int J Gynaecol Obstet. 2007;97:15–20.

    Article  CAS  PubMed  Google Scholar 

  35. Kabyemela E, Goncalves BP, Prevots DR, Morrison R, Harrington W, Gwamaka M, et al. Cytokine profiles at birth predict malaria severity during infancy. PLoS ONE. 2013;8:e77214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med. 2009;361:888–98.

    Article  CAS  PubMed  Google Scholar 

  37. Shulman CE, Dorman EK, Bulmer JN. Malaria as a cause of severe anaemia in pregnancy. Lancet. 2002;360:494.

    Article  CAS  PubMed  Google Scholar 

  38. Ruizendaal E, van Leeuwen E, Mens PF. Peripheral and placental biomarkers in women with placental malaria: a systematic review. Biomark Med. 2015;9:217–39.

    Article  CAS  PubMed  Google Scholar 

  39. Seitz J, Morales-Prieto DM, Favaro RR, Schneider H, Markert UR. Molecular principles of intrauterine growth restriction in Plasmodium falciparum infection. Front Endocrinol (Lausanne). 2019;10:98.

    Article  Google Scholar 

  40. Helegbe GK, Huy NT, Yanagi T, Shuaibu MN, Kikuchi M, Cherif MS, et al. Elevated IL-17 levels in semi-immune anaemic mice infected with Plasmodium berghei ANKA. Malar J. 2018;17:169.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Fitri LE, Sardjono TW, Rahmah Z, Siswanto B, Handono K, Dachlan YP. Low fetal weight is directly caused by sequestration of parasites and indirectly by IL-17 and IL-10 imbalance in the placenta of pregnant mice with malaria. Korean J Parasitol. 2015;53:189–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Nweneka CV, Doherty CP, Cox S, Prentice A. Iron delocalisation in the pathogenesis of malarial anaemia. Trans R Soc Trop Med Hyg. 2010;104:175–84.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We are very grateful to all women who participated in the study. Especial thank extended to all the laboratory and field staff members in Blue Nile State, Sudan. We are also thankful to Ms Inas A. Khalifa from Elrayan Laboratory, who participated in laboratory analysis. This work was supported by Vall d´Hebron Research Institute as part of Samia Omer fellowship of the 4th edition of Science by Women, Foundation Mujeres por África (FMxA), Madrid –Spain.

Author information

Authors and Affiliations

Authors

Contributions

SO, CF, IM, and IA conceived the study and participated in the study coordination. MA conducted the clinical work. MO participated in the immunological laboratory work. SO, IA and AN analyzed and interpreted the data. AN shared the co-drafting of the manuscript. All authors read and approved the final draft of the manuscript.

Corresponding author

Correspondence to Samia Omer.

Ethics declarations

Ethical approval and consent to participate

The study received ethical approval from the Ethical and Scientific Committees of the Tropical Medicine Research Institute, National Centre for Research, and the Directorate of Research, Federal Ministry of Health, Khartoum, Sudan. Written informed consent was obtained from all participants before inclusion in the study.

Conflict of interests

The authors declare that they have no conflict interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Omer, S., Franco-Jarava, C., Noureldien, A. et al. Impact of placental malaria on maternal, placental and fetal cord responses and its role in pregnancy outcomes in women from Blue Nile State, Sudan. Malar J 20, 35 (2021). https://0-doi-org.brum.beds.ac.uk/10.1186/s12936-021-03580-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12936-021-03580-x

Keywords